1
|
Hubbi S, Hao S, Epps J, Ferreri NR. Tumour necrosis factor-alpha at the intersection of renal epithelial and immune cell function. J Physiol 2025. [PMID: 40349332 DOI: 10.1113/jp286756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/10/2025] [Indexed: 05/14/2025] Open
Abstract
This review explores the roles of tumour necrosis factor-alpha (TNF) in kidney physiology and pathology. TNF, produced by renal epithelial cells, regulates glucose, electrolyte, water and urea transport by modulating key transporters such as sodium-glucose co-transporter-2 (SGLT2), sodium-potassium-chloride cotransporter 2 (NKCC2), sodium chloride cotransporter (NCC), epithelial sodium channel (ENaC), aquaporin-2 (AQP2) and urea transporters. Under non-inflammatory conditions, TNF functions as a regulatory 'brake' on water and solute transport, particularly by attenuating NKCC2 and AQP2 activity. Disruption of these actions, coupled with increased salt intake, shifts mice from being salt-resistant to salt-sensitive, thereby altering their blood pressure. In autoimmune diseases, chronic kidney disease (CKD), hypertension with renal inflammation, and sepsis, TNF drives immune responses and disease progression. Although mechanisms underlying tubular epithelial cell (TEC)-immune cell interactions remain unclear, emerging evidence indicates that the spatial organization of immune responses in the kidney is associated with distinct TEC signature phenotypes. Hypertonicity- and NFAT5 (i.e. nuclear factor of activated T cells 5)-driven TNF production in TECs and T lymphocytes may influence immune cell communication by affecting co-stimulatory molecule expression and ENaC activity on macrophages and dendritic cells. Although TNF is generally pathogenic in renal diseases, its inhibition does not always confer protection because its effects on endoplasmic reticulum stress, ion transport, vascular smooth muscle and immune cells are influenced by distinct cellular sources and signalling mechanisms through TNF receptors 1 and 2. Anti-TNF therapies are crucial for treating chronic inflammatory diseases and may also aid in preventing the progression of acute kidney injury to CKD. A more complete understanding of the role of TNF in immunophysiological responses may enable the development of more targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sara Hubbi
- Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Jarred Epps
- Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | | |
Collapse
|
2
|
Hao S, Lasaracina AP, Epps J, Ferreri NR. TNF inhibits NKCC2 phosphorylation by a calcineurin-dependent pathway. Am J Physiol Renal Physiol 2025; 328:F489-F500. [PMID: 40062390 PMCID: PMC12048884 DOI: 10.1152/ajprenal.00251.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/30/2024] [Accepted: 02/26/2025] [Indexed: 03/20/2025] Open
Abstract
We previously demonstrated that tumor necrosis factor-alpha (TNF) inhibits Na+-K+-2Cl- cotransporter (NKCC2) phosphorylation in the thick ascending limb (TAL); however, the underlying mechanism remains unclear. We tested the hypothesis that the induction of calcineurin (CN) activity and the expression of CN isoforms contribute to the mechanism by which TNF inhibits phospho-NKCC2 (pNKCC2) expression. CN activity increased by approximately twofold in primary cultures of medullary (m)TAL cells challenged with mouse recombinant TNF. In contrast, silencing TNF production in mTAL cells using lentivirus U6-TNF-ex4 reduced CN activity. pNKCC2 expression decreased in mTAL cells challenged with TNF, whereas inhibition of CN activity with cyclosporine A (CsA) increased pNKCC2 expression. Although mTAL cells express both the calcineurin A subunit (CNA) α and β isoforms, only CNA β isoform mRNA increased after mTAL cells were challenged with TNF. In vivo, both TNF and CNA β expression increased in outer medulla (OM) from mice given 1% NaCl in the drinking water for 7 days and intrarenal lentivirus silencing of TNF selectively reduced expression of CNA β. Intrarenal injection of a lentivirus that specifically silenced CNA β (U6-CNAβ-ex6) increased pNKCC2 expression and attenuated the inhibitory effects of TNF on pNKCC2 expression in freshly isolated TAL tubules. Collectively, the study is the first to demonstrate that TNF increases CN activity and specifically induces β-isoform expression in the kidney. Since NKCC2 is a known target of the CNA β isoform, these findings suggest that a CN-dependent signaling pathway involving this isoform contributes to the mechanism by which TNF inhibits pNKCC2 expression.NEW & NOTEWORTHY The beneficial immunosuppressive effects of CsA are tempered by renal side effects including reduction of GFR, proximal tubule damage, reduced urinary concentration, fibrosis and hypertension. As chronic administration of CN inhibitors frequently induce hypertension and renal nephropathy in humans, understanding the molecular mechanisms by which CN isoforms regulate the activity of renal transporters may provide the framework for developing new drugs that more selectively modulate the diverse functions of CN.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Anna Pia Lasaracina
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Jarred Epps
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| |
Collapse
|
3
|
Hao S, DelliPizzi A, Lasaracina AP, Ferreri NR. TNF inhibits AQP2 expression via a miR137-dependent pathway. Am J Physiol Renal Physiol 2024; 326:F152-F164. [PMID: 37969102 PMCID: PMC11198993 DOI: 10.1152/ajprenal.00210.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023] Open
Abstract
As miR-137 is a regulator of aquaporin (AQP)2 expression and tumor necrosis factor (TNF) inhibits the expression of several extrarenal AQPs, we tested the hypothesis that TNF inhibits AQP2 in the kidney via a miR-137-dependent mechanism. AQP2 mRNA and protein expression decreased ∼70% and 53%, respectively, in primary renal inner medullary collecting duct (IMCD) cells transfected with a miRNA mimic of mmu-miR-137, suggesting that miR-137 directly targets AQP2 mRNA in these cells. Exposure of IMCD cells for 2 h to 400 mosmol/kgH2O medium increased mmu-miR-137 mRNA expression about twofold, conditions that also increased TNF production approximately fourfold. To determine if the increase in mmu-miR-137 mRNA expression was related to the concomitant increase in TNF, IMCD cells were transfected with a lentivirus construct to silence TNF. This construct decreased mmu-miR-137 mRNA expression by ∼63%, suggesting that TNF upregulates the expression of miR-137. Levels of miR-137 also increased approximately twofold in IMCD tubules isolated from male mice given 1% NaCl in the drinking water for 3 days. Intrarenal lentivirus silencing of TNF increased AQP2 mRNA levels and protein expression concomitant with a decrease in miR-137 levels in tubules isolated from mice given NaCl. The changes in AQP2 expression levels affected the diluting ability of the kidney, which was assessed by measuring urine osmolality and urine volume, as the decrease in these parameters after renal silencing of TNF was prevented on intrarenal administration of miR-137. The study reveals a novel TNF function via a miR-137-dependent mechanism that regulates AQP2 expression and function.NEW & NOTEWORTHY An emerging intratubular tumor necrosis factor system, functioning during normotensive noninflammatory conditions, acts as a breaking mechanism that attenuates both the increases in Na+-K+-2Cl- cotransporter and aquaporin-2 induced by arginine vasopressin, thereby contributing to the regulation of electrolyte balance and blood pressure. A greater appreciation for the role of cytokines as mediators of immunophysiological responses may help reveal the relationship between the immune system and other physiological systems.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - AnnMarie DelliPizzi
- Department of Biology, Dominican University New York, Orangeburg, New York, United States
| | - Anna Pia Lasaracina
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York, United States
| |
Collapse
|
4
|
Crorkin P, Hao S, Ferreri NR. Responses to Ang II (Angiotensin II), Salt Intake, and Lipopolysaccharide Reveal the Diverse Actions of TNF-α (Tumor Necrosis Factor-α) on Blood Pressure and Renal Function. Hypertension 2022; 79:2656-2670. [PMID: 36129177 PMCID: PMC9649876 DOI: 10.1161/hypertensionaha.122.19464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
TNF-α (tumor necrosis factor-alpha) is the best known as a proinflammatory cytokine; yet, this cytokine also has important immunomodulatory and regulatory functions. As the effects of TNF-α on immune system function were being revealed, the spectrum of its activities appeared in conflict with each other before investigators defined the settings and mechanisms by which TNF-α contributed to both host defense and chronic inflammation. These effects reflect self-protective mechanisms that may become harmful when dysregulated. The paradigm of physiological and pathophysiological effects of TNF-α has since been uncovered in the lung, colon, and kidney where its role has been identified in pulmonary edema, electrolyte reabsorption, and blood pressure regulation, respectively. Recent studies on the prohypertensive and inflammatory effects of TNF-α in the cardiovascular system juxtaposed to those related to NaCl and blood pressure homeostasis, the response of the kidney to lipopolysaccharide, and protection against bacterial infections are helping define the mechanisms by which TNF-α modulates distinct functions within the kidney. This review discusses how production of TNF-α by renal epithelial cells may contribute to regulatory mechanisms that not only govern electrolyte excretion and blood pressure homeostasis but also maintain the appropriate local hypersalinity environment needed for optimizing the innate immune response to bacterial infections in the kidney. It is possible that the wide range of effects mediated by TNF-α may be related to severity of disease, amount of inflammation and TNF-α levels, and the specific cell types that produce this cytokine, areas that remain to be investigated further.
Collapse
Affiliation(s)
- Patrick Crorkin
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | - Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, NY
| | | |
Collapse
|
5
|
Watts BA, Tamayo E, Sherwood ER, Good DW. Monophosphoryl lipid A pretreatment suppresses sepsis- and LPS-induced proinflammatory cytokine production in the medullary thick ascending limb. Am J Physiol Renal Physiol 2020; 319:F8-F18. [PMID: 32421349 DOI: 10.1152/ajprenal.00178.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Sepsis is the leading cause of acute kidney injury in critically ill patients. Tumor necrosis factor-α (TNF-α) has been implicated in the pathogenesis of septic kidney injury; however, the sites and mechanisms of renal TNF-α production during sepsis remain to be defined. In the present study, we showed that TNF-α expression is increased in medullary thick ascending limbs (MTALs) of mice with sepsis induced by cecal ligation and puncture. Treatment with lipopolysaccharide (LPS) for 3 h in vitro also increased MTAL TNF-α production. Sepsis and LPS increased MTAL TNF-α expression through activation of the myeloid differentiation factor 88 (MyD88)-IL-1 receptor-associated kinase 1-ERK signaling pathway. Pretreatment with monophosphoryl lipid A (MPLA), a nontoxic immunomodulator that protects against bacterial infection, eliminated the sepsis- and LPS-induced increases in MTAL TNF-α production. The suppressive effect of MPLA on TNF-α was mediated through activation of a phosphatidylinositol 3-kinase-dependent pathway that inhibits MyD88-dependent ERK activation. This likely involves MPLA-phosphatidylinositol 3-kinase-mediated induction of Tollip, which negatively regulates the MyD88-ERK pathway by inhibiting activation of IL-1 receptor-associated kinase 1. These regulatory mechanisms are similar to those previously shown to mediate the effect of MPLA to prevent sepsis-induced inhibition of MTAL [Formula: see text] absorption. These results identify the MTAL as a site of local TNF-α production in the kidney during sepsis and identify molecular mechanisms that can be targeted to attenuate renal TNF-α expression. The ability of MPLA pretreatment to suppress MyD88-dependent ERK signaling in the MTAL during sepsis has the dual beneficial effects of protecting tubule transport functions and attenuating harmful proinflammatory responses.
Collapse
Affiliation(s)
- Bruns A Watts
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Esther Tamayo
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David W Good
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas.,Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
6
|
Hao S, Salzo J, Hao M, Ferreri NR. Regulation of NKCC2B by TNF-α in response to salt restriction. Am J Physiol Renal Physiol 2019; 318:F273-F282. [PMID: 31813248 DOI: 10.1152/ajprenal.00388.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously shown that TNF-α produced by renal epithelial cells inhibits Na+-K+-2Cl- cotransporter (NKCC2) activity as part of a mechanism that attenuates increases in blood pressure in response to high NaCl intake. As the role of TNF-α in the kidney is still being defined, the effects of low salt intake on TNF-α and NKCC2B expression were determined. Mice given a low-salt (0.02% NaCl) diet (LSD) for 7 days exhibited a 62 ± 7.4% decrease in TNF-α mRNA accumulation in the renal cortex. Mice that ingested the LSD also exhibited an ~63% increase in phosphorylated NKCC2 expression in the cortical thick ascending limb of Henle's loop and a concomitant threefold increase in NKCC2B mRNA abundance without a concurrent change in NKCC2A mRNA accumulation. NKCC2B mRNA levels increased fivefold in mice that ingested the LSD and also received an intrarenal injection of a lentivirus construct that specifically silenced TNF-α in the kidney (U6-TNF-ex4) compared with mice injected with control lentivirus. Administration of a single intrarenal injection of murine recombinant TNF-α (5 ng/g body wt) attenuated the increases of NKCC2B mRNA by ~50% and inhibited the increase in phosphorylated NKCC2 by ~54% in the renal cortex of mice given the LSD for 7 days. Renal silencing of TNF-α decreased urine volume and NaCl excretion in mice given the LSD, effects that were reversed when NKCC2B was silenced in the kidney. Collectively, these findings demonstrate that downregulation of renal TNF-α production in response to low-salt conditions contributes to the regulation of NaCl reabsorption via an NKCC2B-dependent mechanism.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Joseph Salzo
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Mary Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
7
|
Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick Ascending Limb Sodium Transport in the Pathogenesis of Hypertension. Physiol Rev 2019; 99:235-309. [PMID: 30354966 DOI: 10.1152/physrev.00055.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb plays a key role in maintaining water and electrolyte balance. The importance of this segment in regulating blood pressure is evidenced by the effect of loop diuretics or local genetic defects on this parameter. Hormones and factors produced by thick ascending limbs have both autocrine and paracrine effects, which can extend prohypertensive signaling to other structures of the nephron. In this review, we discuss the role of the thick ascending limb in the development of hypertension, not as a sole participant, but one that works within the rich biological context of the renal medulla. We first provide an overview of the basic physiology of the segment and the anatomical considerations necessary to understand its relationship with other renal structures. We explore the physiopathological changes in thick ascending limbs occurring in both genetic and induced animal models of hypertension. We then discuss the racial differences and genetic defects that affect blood pressure in humans through changes in thick ascending limb transport rates. Throughout the text, we scrutinize methodologies and discuss the limitations of research techniques that, when overlooked, can lead investigators to make erroneous conclusions. Thus, in addition to advancing an understanding of the basic mechanisms of physiology, the ultimate goal of this work is to understand our research tools, to make better use of them, and to contextualize research data. Future advances in renal hypertension research will require not only collection of new experimental data, but also integration of our current knowledge.
Collapse
Affiliation(s)
| | - Fara Saez
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Casandra M Monzon
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jessica Asirwatham
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| | - Jeffrey L Garvin
- Department of Physiology and Biophysics, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
8
|
Hao S, Hao M, Ferreri NR. Renal-Specific Silencing of TNF (Tumor Necrosis Factor) Unmasks Salt-Dependent Increases in Blood Pressure via an NKCC2A (Na +-K +-2Cl - Cotransporter Isoform A)-Dependent Mechanism. Hypertension 2018; 71:1117-1125. [PMID: 29735631 DOI: 10.1161/hypertensionaha.117.10764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/02/2018] [Accepted: 03/23/2018] [Indexed: 01/11/2023]
Abstract
We tested the hypothesis that TNF (tumor necrosis factor)-α produced within the kidney and acting on the renal tubular system is part of a regulatory mechanism that attenuates increases in blood pressure in response to high salt intake. Intrarenal administration of a lentivirus construct, which specifically silenced TNF in the kidney, did not affect baseline blood pressure. However, blood pressure increased significantly 1 day after mice with intrarenal silencing of TNF ingested 1% NaCl in the drinking water. The increase in blood pressure, which was continuously observed for 11 days, promptly returned to baseline levels when mice were switched from 1% NaCl to tap water. Silencing of renal TNF also increased NKCC2 (Na+-K+-2Cl- cotransporter) phosphorylation and induced a selective increase in NKCC2A (NKCC2 isoform A) mRNA accumulation in both the cortical and medullary thick ascending limb of Henle loop that was neither associated with a compensatory decrease of NKCC2F in the medulla nor NKCC2B in the cortex. The NaCl-mediated increases in blood pressure were completely absent when NKCC2A, using a lentivirus construct that did not alter expression of NKCC2F or NKCC2B, and TNF were concomitantly silenced in the kidney. Moreover, the decrease in urine volume and NaCl excretion induced by renal TNF silencing was abolished when NKCC2A was concurrently silenced, suggesting that this isoform contributes to the transition from a salt-resistant to salt-sensitive phenotype. Collectively, the data are the first to demonstrate a role for TNF produced by the kidney in the modulation of sodium homeostasis and blood pressure regulation.
Collapse
MESH Headings
- Animals
- Blood Pressure/physiology
- Blotting, Western
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay
- Gene Expression Regulation
- Hypertension, Renal/genetics
- Hypertension, Renal/metabolism
- Hypertension, Renal/physiopathology
- Kidney/metabolism
- Kidney/pathology
- Male
- Mice
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Sodium Chloride/toxicity
- Solute Carrier Family 12, Member 1/biosynthesis
- Solute Carrier Family 12, Member 1/genetics
- Transcription, Genetic
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Shoujin Hao
- From the Department of Pharmacology, New York Medical College, Valhalla
| | - Mary Hao
- From the Department of Pharmacology, New York Medical College, Valhalla
| | - Nicholas R Ferreri
- From the Department of Pharmacology, New York Medical College, Valhalla.
| |
Collapse
|
9
|
Graham LA, Dominiczak AF, Ferreri NR. Role of renal transporters and novel regulatory interactions in the TAL that control blood pressure. Physiol Genomics 2017; 49:261-276. [PMID: 28389525 PMCID: PMC5451551 DOI: 10.1152/physiolgenomics.00017.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/27/2017] [Accepted: 04/05/2017] [Indexed: 12/31/2022] Open
Abstract
Hypertension (HTN), a major public health issue is currently the leading factor in the global burden of disease, where associated complications account for 9.4 million deaths worldwide every year. Excessive dietary salt intake is among the environmental factors that contribute to HTN, known as salt sensitivity. The heterogeneity of salt sensitivity and the multiple mechanisms that link high salt intake to increases in blood pressure are of upmost importance for therapeutic application. A continual increase in the kidney's reabsorption of sodium (Na+) relies on sequential actions at various segments along the nephron. When the distal segments of the nephron fail to regulate Na+, the effects on Na+ homeostasis are unfavorable. We propose that the specific nephron region where increased active uptake occurs as a result of variations in Na+ reabsorption is at the thick ascending limb of the loop of Henle (TAL). The purpose of this review is to urge the consideration of the TAL as contributing to the pathophysiology of salt-sensitive HTN. Further research in this area will enable development of a therapeutic application for targeted treatment.
Collapse
Affiliation(s)
- Lesley A Graham
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow Cardiovascular and Medical Sciences, Glasgow, United Kingdom; and
| | - Anna F Dominiczak
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow Cardiovascular and Medical Sciences, Glasgow, United Kingdom; and
| | - Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
10
|
Norlander AE, Saleh MA, Kamat NV, Ko B, Gnecco J, Zhu L, Dale BL, Iwakura Y, Hoover RS, McDonough AA, Madhur MS. Interleukin-17A Regulates Renal Sodium Transporters and Renal Injury in Angiotensin II-Induced Hypertension. Hypertension 2016; 68:167-74. [PMID: 27141060 DOI: 10.1161/hypertensionaha.116.07493] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 04/01/2016] [Indexed: 01/11/2023]
Abstract
Angiotensin II-induced hypertension is associated with an increase in T-cell production of interleukin-17A (IL-17A). Recently, we reported that IL-17A(-/-) mice exhibit blunted hypertension, preserved natriuresis in response to a saline challenge, and decreased renal sodium hydrogen exchanger 3 expression after 2 weeks of angiotensin II infusion compared with wild-type mice. In the current study, we performed renal transporter profiling in mice deficient in IL-17A or the related isoform, IL-17F, after 4 weeks of Ang II infusion, the time when the blood pressure reduction in IL-17A(-/-) mice is most prominent. Deficiency of IL-17A abolished the activation of distal tubule transporters, specifically the sodium-chloride cotransporter and the epithelial sodium channel and protected mice from glomerular and tubular injury. In human proximal tubule (HK-2) cells, IL-17A increased sodium hydrogen exchanger 3 expression through a serum and glucocorticoid-regulated kinase 1-dependent pathway. In mouse distal convoluted tubule cells, IL-17A increased sodium-chloride cotransporter activity in a serum and glucocorticoid-regulated kinase 1/Nedd4-2-dependent pathway. In both cell types, acute treatment with IL-17A induced phosphorylation of serum and glucocorticoid-regulated kinase 1 at serine 78, and treatment with a serum and glucocorticoid-regulated kinase 1 inhibitor blocked the effects of IL-17A on sodium hydrogen exchanger 3 and sodium-chloride cotransporter. Interestingly, both HK-2 and mouse distal convoluted tubule 15 cells produce endogenous IL-17A. IL17F had little or no effect on blood pressure or renal sodium transporter abundance. These studies provide a mechanistic link by which IL-17A modulates renal sodium transport and suggest that IL-17A inhibition may improve renal function in hypertension and other autoimmune disorders.
Collapse
Affiliation(s)
- Allison E Norlander
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Mohamed A Saleh
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Nikhil V Kamat
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Benjamin Ko
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Juan Gnecco
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Linjue Zhu
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Bethany L Dale
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Yoichiro Iwakura
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Robert S Hoover
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Alicia A McDonough
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| | - Meena S Madhur
- From the Departments of Molecular Physiology and Biophysics (A.E.N., B.L.D., M.S.M.) and Microbiology, Immunology, and Pathology (J.G.), Vanderbilt University, Nashville, TN; Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (M.A.S., L.Z., M.S.M.); Faculty of Pharmacy, Department of Pharmacology and Toxicology, Mansoura University, Mansoura, Egypt (M.A.S.); Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles (N.V.K., A.A.M.D.); Department of Medicine, Chicago University School of Medicine, IL (B.K.); Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan (Y.I.); and Division of Renal Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA (R.S.H.)
| |
Collapse
|
11
|
TNF receptors: signaling pathways and contribution to renal dysfunction. Kidney Int 2014; 87:281-96. [PMID: 25140911 DOI: 10.1038/ki.2014.285] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 02/28/2014] [Accepted: 03/06/2014] [Indexed: 12/19/2022]
Abstract
Tumor necrosis factor (TNF), initially reported to induce tumor cell apoptosis and cachexia, is now considered a central mediator of a broad range of biological activities from cell proliferation, cell death and differentiation to induction of inflammation and immune modulation. TNF exerts its biological responses via interaction with two cell surface receptors: TNFR1 and TNFR2. (TNFRs). These receptors trigger shared and distinct signaling pathways upon TNF binding, which in turn result in cellular outputs that may promote tissue injury on one hand but may also induce protective, beneficial responses. Yet the role of TNF and its receptors specifically in renal disease is still not well understood. This review describes the expression of the TNFRs, the signaling pathways induced by them and the biological responses of TNF and its receptors in various animal models of renal diseases, and discusses the current outcomes from use of TNF biologics and TNF biomarkers in renal disorders.
Collapse
|
12
|
Graham LA, Padmanabhan S, Fraser NJ, Kumar S, Bates JM, Raffi HS, Welsh P, Beattie W, Hao S, Leh S, Hultstrom M, Ferreri NR, Dominiczak AF, Graham D, McBride MW. Validation of Uromodulin as a Candidate Gene for Human Essential Hypertension. Hypertension 2014; 63:551-8. [DOI: 10.1161/hypertensionaha.113.01423] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A recent genome-wide association study identified a locus on chromosome 16 in the promoter region of the uromodulin (
UMOD
) gene that is associated with hypertension. Here, we examined the hypertension signal with functional studies in Umod knockout (KO) mice. Systolic blood pressure was significantly lower in KO versus wild-type (WT) mice under basal conditions (KO: 116.6±0.3 mm Hg versus WT: 136.2±0.4 mm Hg;
P
<0.0001). Administration of 2% NaCl did not alter systolic blood pressure in KO mice, whereas it increased in WT mice by ≈33%,
P
<0.001. The average 24-hour urinary sodium excretion in the KO was greater than that of WT mice (
P
<0.001). Chronic renal function curves demonstrate a leftward shift in KO mice, suggesting that the relationship between UMOD and blood pressure is affected by sodium. Creatinine clearance was increased during salt loading with 2% NaCl in the KO mice, leading to augmented filtered Na
+
excretion and further Na
+
loss. The difference in sodium uptake that exists between WT and KO strains was explored at the molecular level. Urinary tumor necrosis factor-α levels were significantly higher in KO mice compared with WT mice (
P
<0.0001). Stimulation of primary thick ascending limb of the loop of Henle cells with exogenous tumor necrosis factor-α caused a reduction in NKCC2A expression (
P
<0.001) with a concurrent rise in the levels of UMOD mRNA (
P
<0.001). Collectively, we demonstrate that UMOD regulates sodium uptake in the thick ascending limb of the loop of Henle by modulating the effect of tumor necrosis factor-α on NKCC2A expression, making UMOD an important determinant of blood pressure control.
Collapse
Affiliation(s)
- Lesley A. Graham
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Sandosh Padmanabhan
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Niall J. Fraser
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Satish Kumar
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - James M. Bates
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Hajamohideen S. Raffi
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Paul Welsh
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Wendy Beattie
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Shoujin Hao
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Sabine Leh
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Michael Hultstrom
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Nicholas R. Ferreri
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Anna F. Dominiczak
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Delyth Graham
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Martin W. McBride
- From the Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom (L.A.G., S.P., N.J.F., P.W., W.B., A.F.D., D.G., M.W.M.); Department of Medicine, University of Oklahoma Health Sciences Centre and Veterans Affairs Medical Center, Oklahoma City (S.K., J.M.B., H.S.R.); Department of Pharmacology, New York Medical College, Valhalla (S.H., N.R.F.); Department of Pathology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
13
|
Ramseyer VD, Garvin JL. Tumor necrosis factor-α: regulation of renal function and blood pressure. Am J Physiol Renal Physiol 2013; 304:F1231-42. [PMID: 23515717 DOI: 10.1152/ajprenal.00557.2012] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Tumor necrosis factor-α (TNF-α) is a pleiotropic cytokine that becomes elevated in chronic inflammatory states such as hypertension and diabetes and has been found to mediate both increases and decreases in blood pressure. High levels of TNF-α decrease blood pressure, whereas moderate increases in TNF-α have been associated with increased NaCl retention and hypertension. The explanation for these disparate effects is not clear but could simply be due to different concentrations of TNF-α within the kidney, the physiological status of the subject, or the type of stimulus initiating the inflammatory response. TNF-α alters renal hemodynamics and nephron transport, affecting both activity and expression of transporters. It also mediates organ damage by stimulating immune cell infiltration and cell death. Here we will summarize the available findings and attempt to provide plausible explanations for such discrepancies.
Collapse
Affiliation(s)
- Vanesa D Ramseyer
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA.
| | | |
Collapse
|
14
|
Hao S, Bellner L, Ferreri NR. NKCC2A and NFAT5 regulate renal TNF production induced by hypertonic NaCl intake. Am J Physiol Renal Physiol 2012; 304:F533-42. [PMID: 23269645 DOI: 10.1152/ajprenal.00243.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Pathways that contribute to TNF production by the kidney are not well defined. Mice given 1% NaCl in the drinking water for 3 days exhibited a 2.5-fold increase in urinary, but not plasma, TNF levels compared with mice given tap water. Since furosemide attenuated the increase in TNF levels, we hypothesized that hypertonic NaCl intake increases renal TNF production by a pathway involving the Na(+)-K(+)-2Cl(-) cotransporter (NKCC2). A 2.5-fold increase in NKCC2A mRNA accumulation was observed in medullary thick ascending limb (mTAL) tubules from mice given 1% NaCl; a concomitant 2-fold increase in nuclear factor of activated T cells 5 (NFAT5) mRNA and protein expression was observed in the outer medulla. Urinary TNF levels were reduced in mice given 1% NaCl after an intrarenal injection of a lentivirus construct designed to specifically knockdown NKCC2A (EGFP-N2A-ex4); plasma levels of TNF did not change after injection of EGFP-N2A-ex4. Intrarenal injection of EGFP-N2A-ex4 also inhibited the increase of NFAT5 mRNA abundance in the outer medulla of mice given 1% NaCl. TNF production by primary cultures of mTAL cells increased approximately sixfold in response to an increase in osmolality to 400 mosmol/kgH2O produced with NaCl and was inhibited in cells transiently transfected with a dnNFAT5 construct. Transduction of cells with EGFP-N2A-ex4 also prevented increases in TNF mRNA and protein production in response to high NaCl concentration and reduced transcriptional activity of a NFAT5 promoter construct. Since NKCC2A expression is restricted to the TAL, NKCC2A-dependent activation of NFAT5 is part of a pathway by which the TAL produces TNF in response to hypertonic NaCl intake.
Collapse
Affiliation(s)
- Shoujin Hao
- Dept. of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | |
Collapse
|
15
|
Battula S, Hao S, Pedraza PL, Stier CT, Ferreri NR. Tumor necrosis factor-alpha induces renal cyclooxygenase-2 expression in response to hypercalcemia. Prostaglandins Other Lipid Mediat 2012; 99:45-50. [PMID: 22800939 DOI: 10.1016/j.prostaglandins.2012.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 06/29/2012] [Accepted: 07/06/2012] [Indexed: 01/01/2023]
Abstract
The effect of tumor necrosis factor-alpha (TNF) on cyclooxygenase-2 (COX-2) expression in the renal outer medulla (OM) was determined in a model of dihydrotachysterol (DHT)-induced hypercalcemia. Increases in serum calcium and water intake were observed during ingestion of a DHT-containing diet in both wild type (WT) and TNF deficient mice (TNF(-/-)). Polyuria and a decrease in body weight were observed in response to DHT treatment in WT and TNF(-/-) mice. A transient elevation in urinary TNF was observed in WT mice treated with DHT. Moreover, increased urinary levels of prostaglandin E(2) (PGE(2)) and a corresponding increase in COX-2 expression in the OM were observed in WT mice fed DHT. Increased COX-2 expression was not observed in TNF(-/-) mice fed DHT, and the characteristics of PGE(2) synthesis were distinct from those in WT mice. This study demonstrates that COX-2 expression in the OM, secondary to hypercalemia, is TNF-dependent.
Collapse
Affiliation(s)
- Sailaja Battula
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | | | | | | | | |
Collapse
|
16
|
Ferreri NR. Going with the flow: regulation of the thick ascending limb. Am J Physiol Renal Physiol 2012; 303:F192-3. [PMID: 22622458 DOI: 10.1152/ajprenal.00258.2012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
17
|
Ferreri NR, Hao S, Pedraza PL, Escalante B, Vio CP. Eicosanoids and tumor necrosis factor-alpha in the kidney. Prostaglandins Other Lipid Mediat 2011; 98:101-6. [PMID: 22101002 DOI: 10.1016/j.prostaglandins.2011.11.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 10/31/2011] [Accepted: 11/03/2011] [Indexed: 12/14/2022]
Abstract
The thick ascending limb of Henle's loop (TAL) is capable of metabolizing arachidonic acid (AA) by cytochrome P450 (CYP450) and cyclooxygenase (COX) pathways and has been identified as a nephron segment that contributes to salt-sensitive hypertension. Previous studies demonstrated a prominent role for CYP450-dependent metabolism of AA to products that inhibited ion transport pathways in the TAL. However, COX-2 is constitutively expressed along all segments of the TAL and is increased in response to diverse stimuli. The ability of Tamm-Horsfall glycoprotein, a selective marker of cortical TAL (cTAL) and medullary (mTAL), to bind TNF and localize it to this nephron segment prompted studies to determine the capacity of mTAL cells to produce TNF and determine its effects on mTAL function. The colocalization of calcium-sensing receptor (CaR) and COX-2 in the TAL supports the notion that activation of CaR induces TNF-dependent COX-2 expression and PGE₂ synthesis in mTAL cells. Additional studies showed that TNF produced by mTAL cells inhibits ⁸⁶Rb uptake, an in vitro correlate of natriuresis, in an autocrine- and COX-2-dependent manner. The molecular mechanism for these effects likely includes inhibition of Na⁺-K⁺-2Cl⁻ cotransporter (NKCC2) expression and trafficking.
Collapse
Affiliation(s)
- Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | | | |
Collapse
|
18
|
Battula S, Hao S, Pedraza PL, Stier CT, Ferreri NR. Tumor necrosis factor-alpha is an endogenous inhibitor of Na+-K+-2Cl- cotransporter (NKCC2) isoform A in the thick ascending limb. Am J Physiol Renal Physiol 2011; 301:F94-100. [PMID: 21511694 DOI: 10.1152/ajprenal.00650.2010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The effects of TNF gene deletion on renal Na(+)-K(+)-2Cl(-) cotransporter (NKCC2) expression and activity were determined. Outer medulla from TNF(-/-) mice exhibited a twofold increase in total NKCC2 protein expression compared with wild-type (WT) mice. This increase was not observed in TNF(-/-) mice treated with recombinant human TNF (hTNF) for 7 days. Administration of hTNF had no effect on total NKCC2 expression in WT mice. A fourfold increase in NKCC2A mRNA accumulation was observed in outer medulla from TNF(-/-) compared with WT mice; NKCC2F and NKCC2B mRNA accumulation was similar between genotypes. The increase in NKCC2A mRNA accumulation was attenuated when TNF(-/-) mice were treated with hTNF. Bumetanide-sensitive O(2) consumption, an in vitro correlate of NKCC2 activity, was 2.8 ± 0.2 nmol·min(-1)·mg(-1) in medullary thick ascending limb tubules from WT, representing ∼40% of total O(2) consumption, whereas, in medullary thick ascending limb tubules from TNF(-/-) mice, it was 5.6 ± 0.3 nmol·min(-1)·mg(-1), representing ∼60% of total O(2) consumption. Administration of hTNF to TNF(-/-) mice restored the bumetanide-sensitive component to ∼30% of total O(2) consumption. Ambient urine osmolality was higher in TNF(-/-) compared with WT mice (2,072 ± 104 vs. 1,696 ± 153 mosmol/kgH(2)O, P < 0.05). The diluting ability of the kidney, assessed by measuring urine osmolality before and after 1 h of water loading also was greater in TNF(-/-) compared with WT mice (174 ± 38 and 465 ± 81 mosmol/kgH(2)O, respectively, P < 0.01). Collectively, these findings suggest that TNF plays a role as an endogenous inhibitor of NKCC2 expression and function.
Collapse
Affiliation(s)
- Sailaja Battula
- Dept. of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | |
Collapse
|
19
|
Hao S, Zhao H, Darzynkiewicz Z, Battula S, Ferreri NR. Differential regulation of NFAT5 by NKCC2 isoforms in medullary thick ascending limb (mTAL) cells. Am J Physiol Renal Physiol 2011; 300:F966-75. [PMID: 21228109 DOI: 10.1152/ajprenal.00408.2010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The effects of Na(+)-K(+)-2Cl(-) cotransporter type 2 (NKCC2) isoforms on the regulation of nuclear factor of activated T cells isoform 5 (NFAT5) were determined in mouse medullary thick ascending limb (mTAL) cells exposed to high NaCl concentration. Primary cultures of mTAL cells and freshly isolated mTAL tubules, both derived from the outer medulla (outer stripe>inner stripe), express NKCC2 isoforms A and F. The relative expression of NKCC2A mRNA was approximately twofold greater than NKCC2F in these preparations. The abundance of NKCC2A mRNA, but not NKCC2F mRNA, increased approximately twofold when mTAL cells were exposed for 2 h to a change in osmolality from 300 to 500 mosmol/kgH₂O, produced with NaCl. Total NKCC2 protein expression also increased. Moreover, a 2.5-fold increase in NFAT5 mRNA accumulation was observed after cells were exposed to 500 mosmol/kgH₂O for 4 h. Laser-scanning cytometry detected a twofold increase in endogenous NFAT5 protein expression in response to high NaCl concentration. Pretreatment with the loop diuretic bumetanide dramatically reduced transcriptional activity of the NFAT5-specific reporter construct TonE-Luc in mTAL cells exposed to high NaCl. Transient transfection of mTAL cells with shRNA vectors targeting NKCC2A prevented increases in NFAT5 mRNA abundance and protein expression and inhibited NFAT5 transcriptional activity in response to hypertonic stress. Silencing of NKCC2F mRNA did not affect NFAT5 mRNA accumulation but partially inhibited NFAT5 transcriptional activity. These findings suggest that NKCC2A and NKCC2F exhibit differential effects on NFAT5 expression and transcriptional activity in response to hypertonicity produced by high NaCl concentration.
Collapse
Affiliation(s)
- Shoujin Hao
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | | | | | | | | |
Collapse
|
20
|
Eng B, Mukhopadhyay S, Vio CP, Pedraza PL, Hao S, Battula S, Sehgal PB, McGiff JC, Ferreri NR. Characterization of a long-term rat mTAL cell line. Am J Physiol Renal Physiol 2007; 293:F1413-22. [PMID: 17670898 DOI: 10.1152/ajprenal.00426.2006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A medullary thick ascending limb (mTAL) cell line, termed raTAL, has been established from freshly isolated rat mTAL tubules and cultured continuously for up to 75 passages; it retains characteristics of mTAL cells even after retrieval from storage in liquid nitrogen for several months. The cells express Tamm-Horsfall glycoprotein (THP), a TAL-specific marker, grow to confluence, exhibit a polygonal morphology characteristic of epithelial cells, and form "domes." Detection of THP, Na(+)-K(+)-2Cl(-) cotransporter (NKCC2), Na(+)-K(+)-ATPase, and renal outer medullary K(+) channel (ROMK) was achieved using indirect immunofluorescence and confocal microscopy. Western blot analysis of NKCC2 expression using two different antibodies revealed a band of approximately 160 kDa, and RT-PCR analysis demonstrated the presence of NKCC2 isoforms A and F, which was confirmed by DNA sequencing; transport of Cl(-) into raTAL cells was inhibited by furosemide. Ouabain- and bumetanide-sensitive oxygen consumption, an index of ion transport activity in the mTAL, was observed in raTAL cells, and the number of domes present was reduced significantly when cells were incubated in the presence of ouabain or bumetanide. The specific activity of Na(+)-K(+)-ATPase activity was determined in raTAL cells (0.67 +/- 0.18 nmol P(i).microg protein(-1).min(-1)), primary cultures of mTAL cells (0.39 +/- 0.08 nmol P(i).microg protein(-1).min(-1)), and freshly isolated mTAL tubules (1.10 +/- 0.29 nmol P(i).microg protein(-1).min(-1)), and approximately 30-50% of total cellular ATPase activity was inhibited by ouabain, in accord with other mTAL preparations. This cell line will be used in studies that address biochemical, molecular, and physiological mechanisms in the mTAL.
Collapse
Affiliation(s)
- Ben Eng
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abdullah HI, Pedraza PL, Hao S, Rodland KD, McGiff JC, Ferreri NR. NFAT regulates calcium-sensing receptor-mediated TNF production. Am J Physiol Renal Physiol 2005; 290:F1110-7. [PMID: 16380462 DOI: 10.1152/ajprenal.00223.2005] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Because nuclear factor of activated T cells (NFAT) has been implicated in TNF production as well as osmoregulation and salt and water homeostasis, we addressed whether calcium-sensing receptor (CaR)-mediated TNF production in medullary thick ascending limb (mTAL) cells was NFAT dependent. TNF production in response to addition of extracellular Ca(2+) (1.2 mM) was abolished in mTAL cells transiently transfected with a dominant-negative CaR construct (R796W) or pretreated with the phosphatidylinositol phospholipase C (PI-PLC) inhibitor U-73122. Cyclosporine A (CsA), an inhibitor of the serine/threonine phosphatase calcineurin, and a peptide ligand, VIVIT, that selectively inhibits calcineurin-NFAT signaling, also prevented CaR-mediated TNF production. Increases in calcineurin activity in cells challenged with Ca(2+) were inhibited after pretreatment with U-73122 and CsA, suggesting that CaR activation increases calcineurin activity in a PI-PLC-dependent manner. Moreover, U-73122, CsA, and VIVIT inhibited CaR-dependent activity of an NFAT construct that drives expression of firefly luciferase in transiently transfected mTAL cells. Collectively, these data verify the role of calcineurin and NFAT in CaR-mediated TNF production by mTAL cells. Activation of the CaR also increased the binding of NFAT to a consensus oligonucleotide, an effect that was blocked by U-73122 and CsA, suggesting that a calcineurin- and NFAT-dependent pathway increases TNF production in mTAL cells. This mechanism likely regulates TNF gene transcription as U-73122, CsA, and VIVIT blocked CaR-dependent activity of a TNF promoter construct. Elucidating CaR-mediated signaling pathways that regulate TNF production in the mTAL will be crucial to understanding mechanisms that regulate extracellular fluid volume and salt balance.
Collapse
|
22
|
Falk R, Hacham M, Nyska A, Foley JF, Domb AJ, Polacheck I. Induction of interleukin-1beta, tumour necrosis factor-alpha and apoptosis in mouse organs by amphotericin B is neutralized by conjugation with arabinogalactan. J Antimicrob Chemother 2005; 55:713-20. [PMID: 15814605 DOI: 10.1093/jac/dki090] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVES To investigate the possibilities that: (i) organ toxicity of amphotericin B-deoxycholate (AMB-DOC) is related to induction of interleukin-1beta (IL-1beta), tumour necrosis factor-alpha (TNF-alpha) and apoptosis in target organs; and (ii) the reduced toxicity resulting from the conjugation of AMB with water-soluble arabinogalactan (AMB-AG), is related to modulation of these parameters. METHODS Organ expression of IL-1beta and TNF-alpha was evaluated by enzyme-linked immunosorbent assay (ELISA) in mouse organ biological fluids and in situ by immunohistochemistry. Tissue damage was evaluated histologically, and apoptosis was demonstrated by terminal dUTP nick end-labelling (TUNEL) staining. AMB-AG conjugate was compared with the micellar (AMB-DOC) and liposomal (AmBisome) AMB formulations. RESULTS Treatment with AMB-AG or AmBisome caused no observable histopathological damage in the kidneys. In contrast, treatment with AMB-DOC resulted in disruptive changes and apoptosis in renal tubular cells. These effects were found to correlate with induction of high levels of IL-1beta and TNF-alpha in kidney lysates. Unlike AMB-AG, AMB-DOC also induced enhanced IL-1beta and TNF-alpha expression in lysates of lungs, brain, liver and spleen. The marked elevation of these inflammation-apoptosis-promoting cytokines after treatment with AMB-DOC may mediate its systemic and local renal damage. Treatment with AMB-AG (but not AmBisome) appears to uniquely modulate the in situ expression of IL-1beta and enhance secretion of TNF-alpha in kidneys, effects possibly involved in prevention of apoptosis. CONCLUSIONS AMB-related toxicity is associated with induction of IL-1beta, TNF-alpha and apoptosis in organs. These effects were not observed with AMB-AG conjugate, suggesting its potential as a safer formulation for therapy.
Collapse
Affiliation(s)
- Rama Falk
- Department of Clinical Microbiology and Infectious Diseases, The Hebrew University-Hadassah Medical Center, PO Box 12000, Jerusalem 91120, Israel
| | | | | | | | | | | |
Collapse
|
23
|
Quan S, Yang L, Shnouda S, Schwartzman ML, Nasjletti A, Goodman AI, Abraham NG. Expression of human heme oxygenase-1 in the thick ascending limb attenuates angiotensin II-mediated increase in oxidative injury. Kidney Int 2004; 65:1628-39. [PMID: 15086901 DOI: 10.1111/j.1523-1755.2004.00562.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. The medullary thick ascending limb of the loop of Henle (TALH) is situated in a site of markedly diminished oxygen tension and, as such, is highly vulnerable to ischemic insult. We hypothesize that selective upregulation of HO-1 in TALH by gene transfer attenuates oxidative stress caused by angiotensin II (Ang II). METHODS An adenoviral vector expressing the human HO-1 under the control of the TALH-specific promoter [Na(+)-K(+)-Cl(-) cotransporter (NKCC2 promoter)] was constructed and the cell specific expression of the recombinant adenovirus was examined using several types of cells, including endothelial, vascular smooth muscle, and TALH cells. The effects of HO-1 transduction on HO-1 expression, HO activity and the response to Ang II with respect to cyclooxygenase-2 (COX-2) up-regulation and oxidative injury [growth-stimulating hormone (GSH) levels and cell death] were determined. RESULTS Western blot and reverse transcription-polymerase chain reaction (RT-PCR) revealed that human HO-1 was selectively expressed in primary cultured TALH cells following infection with Ad-NKCC2-HO-1. In TALH cells infected with Ad-NKCC2-HO-1, Ang II-stimulated prostaglandin E(2) (PGE(2)) levels were reduced by 40%. Ang II caused a marked decrease in GSH levels and this decrease was greatly attenuated in TALH cells transduced with Ad-NKCC2-HO-1. Moreover, Ang II-mediated DNA degradation was completely blocked by the site-specific expression of human HO-1 gene. CONCLUSION These results indicate that TALH cell survival after exposure to oxidative stress injury may be facilitated by selective upregulation of HO-1, thusly blocking inflammation and apoptosis.
Collapse
Affiliation(s)
- Shuo Quan
- Department of Pharmacology, Division of Nephrology, New York Medical College, Valhalla, New York 10595, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Ferreri NR, McGiff JC, Vio CP, Carroll MA. TNFalpha regulates renal COX-2 in the rat thick ascending limb (TAL). Thromb Res 2004; 110:277-80. [PMID: 14592548 DOI: 10.1016/s0049-3848(03)00381-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We have examined cyclooxygenase (COX)-2-dependent mechanisms in preglomerular microvessels and the thick ascending limb (TAL). These renal structures are linchpins in the regulation of the renal circulation and extracellular fluid volume. Cytochrome P450 monooxygenases are the principal oxygenases in the TAL segment; however, COX-2 can be expressed in the TAL, as when challenged by angiotensin II. Glucocorticoids also affect the expression and activity of oxygenases in the TAL. Before adrenalectomy, <2% TAL cells expressed COX-2; after, >30% of TAL cells expressed COX-2. Recruitment of COX-2 is initiated in the renal cortex and proceeds to the medulla associated with: (1) COX-2 mRNA accumulation; (2) increased COX-2 expression; and (3) a two-fold increase in PGE2 production by cortical microsomes. These changes were nullified by dexamethasone. COX-2 mRNA, protein expression and PGE2 synthesis in the TAL are also increased in response to increased extracellular Ca2+. The Ca2+ sensing receptor is G-protein coupled and responds to changes in extracellular Ca2+ concentration by increasing protein kinase C activity to produce expression of COX-2. Thus, multiple signaling pathways contribute to COX-2 expression in TAL cells.
Collapse
Affiliation(s)
- Nicholas R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
25
|
Wei Y, Babilonia E, Pedraza PL, Ferreri NR, Wang WH. Acute application of TNF stimulates apical 70-pS K+ channels in the thick ascending limb of rat kidney. Am J Physiol Renal Physiol 2003; 285:F491-7. [PMID: 12890664 DOI: 10.1152/ajprenal.00104.2003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TNF has been shown to be synthesized by the medullary thick ascending limb (mTAL) (21). In the present study, we used the patch-clamp technique to study the acute effect of TNF on the apical 70-pS K+ channel in the mTAL. Addition of TNF (10 nM) significantly stimulated activity of the 70-pS K+ channel and increased NPo [a product of channel open probability (Po) and channel number (N)] from 0.20 to 0.97. The stimulatory effect of TNF was observed only in cell-attached patches but not in excised patches. Moreover, addition of TNF had no effect on the ROMK-like small-conductance K+ channels in the TAL. The dose-response curve of the TNF effect yielded a Km value of 1 nM, a concentration that increased channel activity to 50% maximal stimulatory effect of TNF. The concentrations required for reaching the plateau of the TNF effect were between 5 and 10 nM. The stimulatory effect of TNF on the 70-pS K+ channel was observed in the presence of N(omega)-nitro-L-arginine methyl ester. This indicated that the effect of TNF was not mediated by a nitric oxide-dependent pathway. Also, inhibition of PKA did not affect the stimulatory effect of TNF. In contrast, inhibition of protein tyrosine kinase not only increased activity of the 70-pS K+ channel but also abolished the effect of TNF. Moreover, inhibition of protein tyrosine phosphatase (PTP) blocked the stimulatory effect of TNF on the 70-pS K+ channel. The notion that the TNF effect results from stimulation of PTP activity is supported by PTP activity assay in which treatment of mTAL cells with TNF significantly increased the activity of PTP. We conclude that TNF stimulates the 70-pS K+ channel via stimulation of PTP in the mTAL.
Collapse
Affiliation(s)
- Yuan Wei
- Dept. of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | |
Collapse
|
26
|
Wang D, Pedraza PL, Abdullah HI, McGiff JC, Ferreri NR. Calcium-sensing receptor-mediated TNF production in medullary thick ascending limb cells. Am J Physiol Renal Physiol 2002; 283:F963-70. [PMID: 12372772 DOI: 10.1152/ajprenal.00108.2002] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Medullary thick ascending limb (mTAL) cells in primary culture express the Ca(2+)-sensing receptor (CaR), a G protein-coupled receptor that senses changes in extracellular Ca(2+) (Ca(o)(2+)) concentration, resulting in increases of intracellular Ca(2+) concentration and PKC activity. Exposure of mTAL cells to either Ca(o)(2+) or the CaR-selective agonist poly-L-arginine increased TNF-alpha synthesis. Moreover, the response to Ca(o)(2+) was enhanced in mTAL cells transfected with a CaR overexpression vector. Transfection of mTAL cells with a TNF promoter construct revealed an increase in reporter gene activity after exposure of the cells to Ca(o)(2+), suggesting that intracellular signaling pathways initiated by means of activation of a CaR contribute to TNF synthesis by a mechanism that involves transcription of the TNF gene. Neutralization of TNF activity with an anti-TNF antibody attenuated Ca(2+)-mediated increases in cyclooxygenase-2 (COX-2) protein expression and PGE(2) synthesis, suggesting that TNF exerts an autocrine effect in the mTAL, which contributes to COX-2-mediated PGE(2) production. Preincubation with the PKC inhibitor bisindolylmaleimide I inhibited Ca(2+)-mediated TNF production. Significant inhibition of COX-2 protein expression and PGE(2) synthesis also was observed when cells were challenged with Ca(o)(2+) in the presence of bisindolylmaleimide I. The data suggest that increases in TNF production subsequent to activation of the CaR may be the basis of an important renal mechanism that regulates salt and water excretion.
Collapse
Affiliation(s)
- Dairong Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | |
Collapse
|
27
|
Todorov V, Müller M, Schweda F, Kurtz A. Tumor necrosis factor-alpha inhibits renin gene expression. Am J Physiol Regul Integr Comp Physiol 2002; 283:R1046-51. [PMID: 12376397 DOI: 10.1152/ajpregu.00142.2002] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Renin, produced in renal juxtaglomerular (JG) cells, is a fundamental regulator of blood pressure. Accumulating evidence suggests that cytokines may directly influence renin production in the JG cells. TNF-alpha, which is one of the key mediators in immunity and inflammation, is known to participate in the control of vascular proliferation and contraction and hence in the pathogenesis of cardiovascular diseases. Thus TNF-alpha may exert its effects on the cardiovascular system through modulation of renal renin synthesis. Therefore we have tested the effect of TNF-alpha on renin transcription in As4.1 cells, which represent transformed mouse JG cells, and in native mouse JG cells in culture. Renin gene expression was also determined in mice lacking the gene for TNF-alpha (TNF-alpha knockout mice). TNF-alpha inhibited renin gene expression via an inhibition of the transcriptional activity, targeting the proximal 4.1 kb of the renin promoter in As4.1 cells. TNF-alpha also attenuated forskolin-stimulated renin gene expression in primary cultures of mouse JG cells. Mice lacking the TNF-alpha gene had almost threefold higher basal renal renin mRNA abundance relative to the control strain. The general physiological regulation of renin expression by salt was not disturbed in TNF-alpha knockout mice. Our data suggest that TNF-alpha inhibits renin gene transcription at the cellular level and thus may act as a modulator of renin synthesis in (physio)pathological situations.
Collapse
Affiliation(s)
- Vladimir Todorov
- Institut für Physiologie I, Universität Regensburg, D-93040 Regensburg, Germany.
| | | | | | | |
Collapse
|
28
|
Schumacher M, Frey FJ, Montani JP, Dick B, Frey BM, Ferrari P. Salt-sensitivity of blood pressure and decreased 11beta-hydroxysteroid dehydrogenase type 2 activity after renal transplantation. Transplantation 2002; 74:66-72. [PMID: 12134101 DOI: 10.1097/00007890-200207150-00012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND High blood pressure (BP) predicts a poor long-term kidney graft outcome. The mechanisms for hypertension in renal graft recipients are only partly understood. There is evidence that BP is salt dependent in renal transplant recipients. We hypothesize that renal transplantation induces salt sensitivity by decreasing 11beta-hydroxysteroid dehydrogenase type 2 (11betaHSD2) activity. METHODS A syngenic uninephrectomized rat transplantation model (Lewis to Lewis) (n=7) was used to demonstrate salt sensitivity after transplantation. Sham-operated (n=5) and denervated rats (n=5) were used as controls. In all rats, BP was measured continuously by telemetry 24 hr a day, whereas the rats were set successively on a normal- (0.45% NaCl), high- (8% NaCl), low- (0.1% NaCl), and, again, normal-salt (0.45% NaCl) diet during a 6-day period to assess salt-related changes in mean arterial pressure (MAP). 11betaHSD2 activity was assessed by determining the ratio of corticosterone to dehydrocorticosterone metabolites (THB+5alphaTHB)/THA in urine. RESULTS After uninephrectomy and implantation of the telemetry device, MAP was comparable in rats assigned to undergo sham operation (100+/-3 mmHg), denervation (105+/-5 mmHg), or transplantation (102+/-6 mmHg). When animals were switched from the normal- to high-salt diet, the increase in MAP was more pronounced in the transplanted group (13.9+/-5.1 mmHg) than in those undergoing sham operation (5.1+/-1.7 mmHg, P<0.004) or denervation (7.1+/-1.8 mmHg, P<0.021). Urinary (THB+5alphaTHB)/THA increased more than 2-fold in the transplanted rats but remained stable in the sham-operated and denervated animals (P<0.0001), indicating reduced activity of 11betaHSD2. CONCLUSION Syngenic renal transplantation causes salt sensitivity with increased BP associated with a reduced activity of 11betaHSD2.
Collapse
Affiliation(s)
- Martin Schumacher
- Division of Nephrology and Hypertension and Institute of Physiology, University of Fribourg, Switzerland
| | | | | | | | | | | |
Collapse
|
29
|
Wang D, An SJ, Wang WH, McGiff JC, Ferreri NR. CaR-mediated COX-2 expression in primary cultured mTAL cells. Am J Physiol Renal Physiol 2001; 281:F658-64. [PMID: 11553512 DOI: 10.1152/ajprenal.2001.281.4.f658] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Primary cultures of medullary thick ascending limb (mTAL) cells retain the capacity to express calcium-sensing receptor (CaR) mRNA and protein. Increases in cyclooxygenase-2 (COX-2) mRNA accumulation, protein expression, and PGE(2) synthesis were observed in a dose- and time-dependent manner after exposure of these cells to extracellular calcium (Ca(o)(2+)). Moreover, transfection of mTAL cells with a CaR overexpression vector significantly enhanced COX-2 expression and PGE(2) production in response to calcium compared with cells transfected with an empty vector. Challenge with the CaR-selective agonist poly-L-arginine (PLA) also increased COX-2 mRNA accumulation, protein expression, and PGE(2) synthesis. Furthermore, Ca(o)(2+)- and PLA-mediated PGE(2) production was abolished in the presence of NS-398 or nimesulide, two different COX-2-selective inhibitors. These data suggest that intracellular signaling mechanisms initiated via activation of CaR contribute to COX-2-dependent PGE(2) synthesis in the mTAL. Because Ca(o)(2+) concentration varies along Henle's loop, calcium may contribute to salt and water balance via a COX-2- and CaR-dependent mechanism. Thus novel calcimimetics might be useful in conditions such as hypertension in which manipulation of extracellular fluid volume provides beneficial effects.
Collapse
Affiliation(s)
- D Wang
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | |
Collapse
|
30
|
Odeh M. The role of tumour necrosis factor-alpha in the pathogenesis of complicated falciparum malaria. Cytokine 2001; 14:11-8. [PMID: 11298488 DOI: 10.1006/cyto.2001.0845] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Plasmodium falciparum malaria is the most important parasitic infection of humans and is one of the most serious health problems facing the inhabitants of developing countries. It is responsible for about 2 million deaths every year. To date there is no specific treatment for the disease apart from anti-malarials. The declining sensitivity to these drugs is a serious therapeutic problem, while no safe and effective vaccine is likely to be available for general use in the near future. There is now abundant laboratory and clinical evidence to suggest that tumour necrosis factor-alpha (TNF-alpha) plays a major role in the pathogenesis of complicated falciparum malaria. Modulation of TNF-alpha response in combination with the current anti-malarial drugs, may represent a novel approach to the treatment of the serious complications associated with the disease.
Collapse
Affiliation(s)
- M Odeh
- Department of Internal Medicine, Bnai Zion Medical Center, Haifa, Israel
| |
Collapse
|
31
|
Askari B, Ferreri NR. Regulation of prostacyclin synthesis by angiotensin II and TNF-alpha in vascular smooth muscle. Prostaglandins Other Lipid Mediat 2001; 63:175-87. [PMID: 11305695 DOI: 10.1016/s0090-6980(01)00098-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We had previously established that in a model of Ang II-induced hypertension, administration of an anti-TNF-alpha antibody caused additional increases in mean arterial pressure. Production of vasodilator prostanoids (i.e. PGI2 and PGE2) is increased by Ang II in vascular smooth muscle and is part of a counter-regulatory mechanism that opposes increases in vascular tone. We, therefore, examined the effects of TNF-alpha on Ang II-induced increases in PGI2 production in vascular smooth muscle cells (VSMC). Addition of Ang II caused an increase in the production of PGI2, while addition of TNF-alpha had no effect. However, pretreatment with TNF-alpha potentiated the stimulatory effects of Ang II. The potentiating effect of TNF-alpha was neither at the level of prostacyclin synthetase nor at the level of acyl hydrolase activity. This potentiation was dependent on tyrosine kinase activity, as preincubation with genistein completely abolished the effect of TNF-alpha. TNF-alpha upregulated AA-induced PGI2 synthesis, indicating that the effect of TNF-alpha is at the level of cyclooxygenase (COX). These data suggest that TNF-alpha potentiates Ang II-induced synthesis of PGI2 and PGE2 in a tyrosine kinase-dependent manner, an effect that may contribute to the counter-regulatory influence of prostaglandins on the pressor effects of Ang II in the vasculature.
Collapse
Affiliation(s)
- B Askari
- Department of Pathology, University of Washington School of Medicine, Seattle 98125, USA
| | | |
Collapse
|
32
|
Abstract
TNF-alpha has been found in the retina. Hyperoxia and hypoxia regulate TNF-alpha expression. TNF-alpha is an important factor in inflammation and angiogenesis. Dexamethasone inhibits TNF-alpha production. Changes in TNF-alpha expression in the retina may play an important role in the development of oxygen-induced retinopathy. Oxygen-induced retinopathy was produced in C57BL6 mice by exposure to 75% oxygen at Postnatal Day 7 (P7) for 5 days and the mice recovered in room air until Day 17 (P17). Dexamethasone was administered at 0.5 mg/kg/day once daily subcutaneously during the 5 days of oxygen exposure. TNF-alpha expression was evaluated at Day 7 prior to oxygen exposure, at Day 12 (P12) immediately upon removal from oxygen, and at Day 17, the time of maximal vasoproliferation by RT-PCR. TNF-alpha is developmentally regulated in the retinae of C57BL6 mice. From P7 to P12, there is a 3-fold increase in TNF-alpha expression and from P7 to P17 there is a 2.7-fold increase. There was 2.7-fold suppression in expression immediately following oxygen exposure at P12. The expression was dramatically increased at P17, the time of maximal vasoproliferation. Dexamethasone inhibited the expression of TNF-alpha at P17 by 6.4-fold. At this dose, it also suppressed the baseline TNF-alpha expression in the mouse model. In summary, TNF-alpha is altered in the development of oxygen-induced retinopathy in the mouse. It increased markedly during the vasoproliferative phase and was suppressed by dexamethasone. Modulation of TNF-alpha expression may provide a potential site of action for future therapeutic targets.
Collapse
Affiliation(s)
- P Yossuck
- Department of Pediatrics, Division of Neonatology, Georgetown University Children's Medical Center, Washington, DC 20007 , USA
| | | | | | | |
Collapse
|
33
|
Croft KD, McGiff JC, Sanchez-Mendoza A, Carroll MA. Angiotensin II releases 20-HETE from rat renal microvessels. Am J Physiol Renal Physiol 2000; 279:F544-51. [PMID: 10966934 DOI: 10.1152/ajprenal.2000.279.3.f544] [Citation(s) in RCA: 106] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We studied hydroxyeicosatetraenoic acid (HETE) release in response to ANG II from preglomerular microvessels (PGMVs), the vascular segment governing changes in renal vascular resistance. PGMVs were isolated from Sprague-Dawley rats and incubated with NADPH and hormones at 37 degrees C. Eicosanoids were extracted, and cytochrome P-450 (CYP)-derived HETEs were purified and quantitated by negative chemical ionization gas chromatography-mass spectroscopy. PGMVs produced primarily 20- and 19-HETEs, namely, 7.9 +/- 1.7 and 2.2 +/- 0.5 ng/mg protein, respectively. ANG II (5 nM) increased CYP-HETE release by two- to threefold; bradykinin, phenylephrine, and Ca(2+) ionophore were without effect. [Sar(1)]ANG II (0.1-100 microM) dose dependently stimulated 19- and 20-HETEs, an effect blocked by the AT(2)-receptor antagonist PD-123319 as well as by U-73122, a phospholipase C inhibitor. Microvascular 20-HETE release was increased more than twofold by the third day in response to ANG II (120 ng. kg(-1). min(-1)) infused subcutaneously for 2 wk; it was not further enhanced after 14 days, although blood pressure continued to rise. Thus an AT(2)-phospholipse C effector unit is associated with synthesis of a vasoconstrictor product, 20-HETE, in a key renovascular segment.
Collapse
Affiliation(s)
- K D Croft
- Department of Medicine, University of Western Australia, Perth WA 6847, Australia
| | | | | | | |
Collapse
|
34
|
Ferreri NR, An SJ, McGiff JC. Cyclooxygenase-2 expression and function in the medullary thick ascending limb. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:F360-8. [PMID: 10484519 DOI: 10.1152/ajprenal.1999.277.3.f360] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The medullary thick ascending limb (MTAL) metabolizes arachidonic acid (AA) via cytochrome P-450 (CyP450)- and cyclooxygenase (COX)-dependent pathways. In the present study, we demonstrated that the COX-2-selective inhibitor, NS-398, prevented tumor necrosis factor-alpha (TNF)- and phorbol myristate acetate (PMA)-mediated increases in PGE(2) production by cultured MTAL cells. Accumulation of COX-2, but not COX-1, mRNA increased when cells were challenged with TNF (1 nM) or PMA (1 microM). Pretreatment of cells for 30 min with actinomycin D (AcD, 1 microM) had little effect on COX-2 mRNA accumulation in unstimulated cells or in cells challenged with either TNF or PMA. Moreover, a posttranscriptional mechanism(s) appears to contribute significantly to COX-2 mRNA accumulation as pretreatment for 15 min with cycloheximide (CHX, 1 microM) caused a superinduction of COX-2 mRNA accumulation in unstimulated cells as well as in cells challenged with either TNF or PMA. Expression of COX-2 protein in unstimulated MTAL cells was attenuated by preincubation for 2 h with dexamethasone (Dex, 2 microM); however, Dex had little or no effect on COX-2 expression in cells challenged with either PMA or TNF. The time-dependent inhibition of 86Rb uptake by MTAL cells challenged with TNF was diminished by pretreating cells with NS-398. These data suggest that TNF-mediated induction of COX-2 protein expression accounted for the lag-time required for this cytokine to inhibit 86Rb uptake in MTAL cells.
Collapse
Affiliation(s)
- N R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA.
| | | | | |
Collapse
|
35
|
McGiff JC, Quilley J. 20-HETE and the kidney: resolution of old problems and new beginnings. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:R607-23. [PMID: 10484476 DOI: 10.1152/ajpregu.1999.277.3.r607] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The protean properties of 20-hydroxyeicosatetraenoic acid (HETE), vasoactivity, mitogenicity, and modulation of transport in key nephron segments, serve as the basis for the essential roles of 20-HETE in the regulation of the renal circulation and electrolyte excretion and as a second messenger for endothelin-1 and mediator of selective renal effects of ANG II. Renal autoregulation and tubular glomerular feedback are mediated by 20-HETE through constriction of preglomerular arterioles, responses that are maintained by 20-HETE inhibition of calcium-activated potassium channels. 20-HETE modulates ion transport in the proximal tubules and the thick ascending limb by affecting the activities of Na+-K+-ATPase and the Na+-K+-2Cl- cotransporter, respectively. The range and diversity of activity of 20-HETE derives in large measure from COX-dependent transformation of 20-HETE to products affecting vasomotion and salt and water excretion. Nitric oxide (NO) exerts a negative modulatory effect on 20-HETE formation; inhibition of NO synthesis produces marked perturbation of renal function resulting from increased 20-HETE production. 20-HETE is an essential component of interactions involving several hormonal systems that have central roles in blood pressure homeostasis, including angiotensins, endothelins, NO, and cytokines. 20-HETE is the preeminent renal eicosanoid, overshadowing PGE2 and PGI2. This review is intended to provide evidence for the physiological roles for cytochrome P-450-derived eicosanoids, particularly 20-HETE, and seeks to extend this knowledge to a conceptual framework for overall cardiovascular function.
Collapse
Affiliation(s)
- J C McGiff
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA.
| | | |
Collapse
|
36
|
Yanagisawa H, Nodera M, Umemori Y, Shimoguchi Y, Wada O. Role of angiotensin II, endothelin-1, and nitric oxide in HgCl2-induced acute renal failure. Toxicol Appl Pharmacol 1998; 152:315-26. [PMID: 9853001 DOI: 10.1006/taap.1998.8459] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To elucidate the mechanisms underlying the development of HgCl2-induced acute renal failure (ARF), we examined the expression of endothelin (ET)-1, endothelial (e) nitric oxide synthase (NOS) and inducible (i) NOS, and a role of angiotensin II (ANG II) and tumor necrosis factor (TNF) in glomeruli and cortices from rats at 20 h after exposure of HgCl2. Prepro-ET-1 and iNOS mRNA were significantly increased in glomeruli and cortices from rats with HgCl2-induced ARF. However, eNOS mRNA was markedly decreased in glomeruli of rats with HgCl2-induced ARF. Blockade of the action of endogenous ANG II with TCV-116, an ANG II receptor type 1 antagonist, or prior administration of TNF antibody (Ab) neutralizing TNF bioactivity or aminoguanidine, an iNOS inhibitor, substantially suppressed the increase in the expression of prepro-ET-1 or iNOS mRNA seen in rats with HgCl2-induced ARF. Both TCV-116 and TNF Ab had no effects on the expression of eNOS mRNA. The abundance of ET-1, iNOS, and eNOS proteins was paralleled by the magnitude of each mRNA expression. Additionally, the aggravation of blood urea nitrogen and serum Cr observed in rats with HgCl2-induced ARF were significantly ameliorated together with the alleviation of proximal tubule epithelial cell injury when the expression of prepro-ET-1 or iNOS mRNA was blunted by prior administration of TCV-116 or prior injection of TNF Ab or aminoguanidine. These observations indicate that ANG II, ET-1, and NO may play an important role in the progression of HgCl2-induced ARF through the acceleration of proximal tubule epithelial cell injury and the deterioration of glomerular hemodynamics. In HgCl2-induced ARF, the gene expression of ET-1 or iNOS is at least in part up-regulated at the transcription level by endogenous ANG II or TNF.
Collapse
Affiliation(s)
- H Yanagisawa
- Department of Hygiene and Preventive Medicine, Faculty of Medicine, Saitama Medical School, Japan
| | | | | | | | | |
Collapse
|
37
|
McGiff JC, Ferreri NR, Escalante BA, Carroll MA. Interactions of renal cytochrome P450 (CYP), angiotensin (AII) and tumor necrosis factor-alpha (TNF): implications for ion transport. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1998; 433:103-7. [PMID: 9561114 DOI: 10.1007/978-1-4899-1810-9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- J C McGiff
- New York Medical College, Department of Pharmacology, Valhalla 10595, USA
| | | | | | | |
Collapse
|
38
|
Rui-Mei L, Kara AU, Sinniah R. Dysregulation of cytokine expression in tubulointerstitial nephritis associated with murine malaria. Kidney Int 1998; 53:845-52. [PMID: 9551390 DOI: 10.1111/j.1523-1755.1998.00848.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We examined the circulating levels of the proinflammatory cytokines tumor necrosis factor-alpha (TNF-alpha), interleukin (IL)-1 alpha, IL-6, granulocyte macrophage-colony stimulating factor (GM-CSF), and the anti-inflammatory cytokine IL-10, and their expression in kidneys acutely infected with murine malaria parasite P. berghei ANKA in C57BL/6J mice. Groups of six mice sacrificed on days 5, 10, 15, and 20, and normal controls were used for cytokine analysis. High concentrations of TNF-alpha and IL-10 were detected in plasma as shown by ELISA, and elevated levels of mRNA specific for TNF-alpha and IL-10 in infected kidneys were demonstrated by reverse transcription-polymerase chain reaction (RT-PCR) analysis. Kidney sections stained with antibodies against TNF-alpha, IL-1 alpha, IL-6, GM-CSF and IL-10 for immunohistochemistry showed markedly enhanced staining for TNF-alpha, and progressively increased staining for IL-1 alpha and IL-6 both in the tubules and the walls of arteries during the course of infection. The endothelia of blood vessels and inflammatory cells located around small arteries showed positive staining for GM-CSF from day 10 onwards. Unlike the staining for proinflammatory cytokines, the anti-inflammatory cytokine IL-10 showed strongly positive staining in normal tubules and walls of arteries, especially in the brush border of proximal tubules, but the staining intensity decreased dramatically after day 15 post-infection. A strongly positive correlation was found between the antibody staining for TNF-alpha/IL-1 alpha in tubules, and the severity of proteinuria. In contrast, there was an inverse correlation between the staining for IL-10 with TNF-alpha/IL-1 alpha, and the degree of proteinuria. Plenty of pigmented macrophages showed positive staining both for proinflammatory and anti-inflammatory cytokines in the tubulointerstitium. Our findings imply that the up-regulation of proinflammatory cytokines and the dysregulation of anti-inflammatory cytokines are involved in the pathogenesis of tubulointerstitial nephritis associated with malaria.
Collapse
Affiliation(s)
- L Rui-Mei
- Department of Pathology, National University of Singapore, Singapore
| | | | | |
Collapse
|
39
|
Ferreri NR, Escalante BA, Zhao Y, An SJ, McGiff JC. Angiotensin II induces TNF production by the thick ascending limb: functional implications. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:F148-55. [PMID: 9458834 DOI: 10.1152/ajprenal.1998.274.1.f148] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The effects of angiotensin II (ANG II) on tumor necrosis factor-alpha (TNF) production were determined in freshly isolated tubules from the medullary thick ascending limb (MTAL). ANG II (10(-9) M) increased the accumulation of TNF mRNA associated with enhanced production of TNF by approximately five- to sixfold. ANG II also increased prostaglandin E2 (PGE2) production by the MTAL in a dose-dependent manner and exerted biphasic differential effects on 86Rb uptake, depending on the exposure time of the tubules to the peptide and the doses used. Low-dose ANG II (10(-11) M) increased 86Rb uptake by MTAL tubules after a "short-term" (15 min) challenge, whereas uptake was inhibited after a "long-term" (3 h) incubation period. High-dose ANG II (10(-6) M) inhibited MTAL 86Rb uptake, irrespective of incubation time. Uptake of 86Rb was inhibited by approximately 60% in MTAL tubules that were challenged for 3 h with ANG II. The inhibitory action of ANG II was prevented by eliminating the participation of either TNF with antisera to the cytokine or PGE2 by inhibition of cyclooxygenase with indomethacin. We conclude that ANG II regulates TNF production in the MTAL, an interaction that affects 86Rb uptake via an eicosanoid-dependent mechanism in this nephron segment.
Collapse
Affiliation(s)
- N R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla 10595, USA
| | | | | | | | | |
Collapse
|
40
|
Ferreri NR, Zhao Y, Takizawa H, McGiff JC. Tumor necrosis factor-alpha-angiotensin interactions and regulation of blood pressure. J Hypertens 1997; 15:1481-4. [PMID: 9431855 DOI: 10.1097/00004872-199715120-00016] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To compare the levels of tumor necrosis factor-alpha (TNF) produced by medullary thick ascending limb tubules (MTAL) obtained from normotensive and angiotensin II (Ang II)-dependent hypertensive rats and determine whether TNF participates in a mechanism that opposes elevation of blood pressure by Ang II. DESIGN We have previously demonstrated that in-vitro administration of Ang II increases production of TNF and prostaglandin E2 (PGE2) by the MTAL. We hypothesize that production of TNF and PGE2 by the MTAL is elevated in in-vivo models of Ang II-dependent hypertension and acts to modulate the pressor effects of Ang II. Thus, inhibition of TNF should disclose whether this cytokine acts to modulate Ang II-induced hypertension. METHODS MTAL tubules obtained from normotensive and Ang II-dependent hypertensive rats were isolated by enzymatic digestion and sieving. Tubules were cultured in the absence of exogenous Ang II. TNF and PGE2 levels were measured by enzyme-linked immunosorbent assay. Anti-TNF antiserum was administered intravenously to normotensive and Ang II-dependent hypertensive rats and their mean arterial pressures were measured. RESULTS Production of TNF and PGE2 was significantly greater in MTAL tubules isolated from Ang II hypertensive rats than it was in those from normotensive controls. Administration of anti-TNF antiserum exacerbated the Ang II-mediated increase in mean arterial pressure. CONCLUSIONS The higher levels of production of TNF and PGE2 by MTAL tubules isolated from Ang II hypertensive rats compared with those of normotensive controls are consistent with results of in-vitro experiments showing that administration of Ang II increases production of TNF and PGE2 by the MTAL. TNF and PGE2 participate in a counter-regulatory mechanism that opposes the pressor actions of Ang II.
Collapse
Affiliation(s)
- N R Ferreri
- Department of Pharmacology, New York Medical College, Valhalla 10595, USA
| | | | | | | |
Collapse
|
41
|
KANETO H, MORRISSEY JJ, McCRACKEN R, ISHIDOYA S, REYES AA, KLAHR S. The expression of mRNA for tumour necrosis factor-? increases in the obstructed kidney of rats soon after unilateral ureteral ligation. Nephrology (Carlton) 1996. [DOI: 10.1111/j.1440-1797.1996.tb00082.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
|
43
|
Beeri R, Symon Z, Brezis M, Ben-Sasson SA, Baehr PH, Rosen S, Zager RA. Rapid DNA fragmentation from hypoxia along the thick ascending limb of rat kidneys. Kidney Int 1995; 47:1806-10. [PMID: 7543962 DOI: 10.1038/ki.1995.249] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Extensive DNA fragmentation, a marker for programmed cell death, was selectively and rapidly induced by hypoxia in the thick ascending limbs of rat kidneys. In isolated perfused kidneys, DNA breaks were present in medullary tubules as early as after 10 minutes of local hypoxia and were prevented by reduction of metabolic work. In a model of radiocontrast-induced acute renal failure, DNA breaks were detected selectively along thick ascending limbs as early as 15 minutes following insult, preceding overt morphological damage. Hypoxia induces rapid DNA fragmentation along thick ascending limbs, where programmed cell death could play an important role in nephron injury and kidney failure.
Collapse
Affiliation(s)
- R Beeri
- Department of Medicine, Hadassah University Hospital-Mt. Scopus, Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|